Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nature ; 615(7953): 705-711, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922598

RESUMEN

Artificial sweeteners are used as calorie-free sugar substitutes in many food products and their consumption has increased substantially over the past years1. Although generally regarded as safe, some concerns have been raised about the long-term safety of the consumption of certain sweeteners2-5. In this study, we show that the intake of high doses of sucralose in mice results in immunomodulatory effects by limiting T cell proliferation and T cell differentiation. Mechanistically, sucralose affects the membrane order of T cells, accompanied by a reduced efficiency of T cell receptor signalling and intracellular calcium mobilization. Mice given sucralose show decreased CD8+ T cell antigen-specific responses in subcutaneous cancer models and bacterial infection models, and reduced T cell function in models of T cell-mediated autoimmunity. Overall, these findings suggest that a high intake of sucralose can dampen T cell-mediated responses, an effect that could be used in therapy to mitigate T cell-dependent autoimmune disorders.


Asunto(s)
Sacarosa , Edulcorantes , Linfocitos T , Animales , Ratones , Sacarosa/análogos & derivados , Edulcorantes/administración & dosificación , Edulcorantes/efectos adversos , Edulcorantes/farmacología , Edulcorantes/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología , Inocuidad de los Alimentos , Señalización del Calcio/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/inmunología , Infecciones Bacterianas/inmunología , Neoplasias/inmunología , Autoinmunidad/efectos de los fármacos , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología
2.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23746840

RESUMEN

A "switch" from oxidative phosphorylation (OXPHOS) to aerobic glycolysis is a hallmark of T cell activation and is thought to be required to meet the metabolic demands of proliferation. However, why proliferating cells adopt this less efficient metabolism, especially in an oxygen-replete environment, remains incompletely understood. We show here that aerobic glycolysis is specifically required for effector function in T cells but that this pathway is not necessary for proliferation or survival. When activated T cells are provided with costimulation and growth factors but are blocked from engaging glycolysis, their ability to produce IFN-γ is markedly compromised. This defect is translational and is regulated by the binding of the glycolysis enzyme GAPDH to AU-rich elements within the 3' UTR of IFN-γ mRNA. GAPDH, by engaging/disengaging glycolysis and through fluctuations in its expression, controls effector cytokine production. Thus, aerobic glycolysis is a metabolically regulated signaling mechanism needed to control cellular function.


Asunto(s)
Glucólisis , Activación de Linfocitos , Fosforilación Oxidativa , Linfocitos T/citología , Linfocitos T/metabolismo , Regiones no Traducidas 3' , Animales , Proliferación Celular , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Interferón gamma/genética , Listeria monocytogenes , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Linfocitos T/inmunología
3.
Nat Immunol ; 15(4): 323-32, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24562310

RESUMEN

The ligation of Toll-like receptors (TLRs) leads to rapid activation of dendritic cells (DCs). However, the metabolic requirements that support this process remain poorly defined. We found that DC glycolytic flux increased within minutes of exposure to TLR agonists and that this served an essential role in supporting the de novo synthesis of fatty acids for the expansion of the endoplasmic reticulum and Golgi required for the production and secretion of proteins that are integral to DC activation. Signaling via the kinases TBK1, IKKɛ and Akt was essential for the TLR-induced increase in glycolysis by promoting the association of the glycolytic enzyme HK-II with mitochondria. In summary, we identified the rapid induction of glycolysis as an integral component of TLR signaling that is essential for the anabolic demands of the activation and function of DCs.


Asunto(s)
Células Dendríticas/inmunología , Glucólisis , Quinasa I-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Ácidos Grasos/biosíntesis , Glucólisis/efectos de los fármacos , Glucólisis/genética , Glucólisis/inmunología , Hexoquinasa/metabolismo , Quinasa I-kappa B/genética , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptores Toll-Like/agonistas
4.
Immunity ; 42(1): 41-54, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25607458

RESUMEN

Naive T cells undergo metabolic reprogramming to support the increased energetic and biosynthetic demands of effector T cell function. However, how nutrient availability influences T cell metabolism and function remains poorly understood. Here we report plasticity in effector T cell metabolism in response to changing nutrient availability. Activated T cells were found to possess a glucose-sensitive metabolic checkpoint controlled by the energy sensor AMP-activated protein kinase (AMPK) that regulated mRNA translation and glutamine-dependent mitochondrial metabolism to maintain T cell bioenergetics and viability. T cells lacking AMPKα1 displayed reduced mitochondrial bioenergetics and cellular ATP in response to glucose limitation in vitro or pathogenic challenge in vivo. Finally, we demonstrated that AMPKα1 is essential for T helper 1 (Th1) and Th17 cell development and primary T cell responses to viral and bacterial infections in vivo. Our data highlight AMPK-dependent regulation of metabolic homeostasis as a key regulator of T cell-mediated adaptive immunity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Adaptación Fisiológica/inmunología , Animales , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Metabolismo Energético , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Inmunomodulación , Activación de Linfocitos/genética , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Biosíntesis de Proteínas/genética
6.
Nature ; 544(7650): 372-376, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28425994

RESUMEN

The non-essential amino acids serine and glycine are used in multiple anabolic processes that support cancer cell growth and proliferation (reviewed in ref. 1). While some cancer cells upregulate de novo serine synthesis, many others rely on exogenous serine for optimal growth. Restriction of dietary serine and glycine can reduce tumour growth in xenograft and allograft models. Here we show that this observation translates into more clinically relevant autochthonous tumours in genetically engineered mouse models of intestinal cancer (driven by Apc inactivation) or lymphoma (driven by Myc activation). The increased survival following dietary restriction of serine and glycine in these models was further improved by antagonizing the anti-oxidant response. Disruption of mitochondrial oxidative phosphorylation (using biguanides) led to a complex response that could improve or impede the anti-tumour effect of serine and glycine starvation. Notably, Kras-driven mouse models of pancreatic and intestinal cancers were less responsive to depletion of serine and glycine, reflecting an ability of activated Kras to increase the expression of enzymes that are part of the serine synthesis pathway and thus promote de novo serine synthesis.


Asunto(s)
Glicina/deficiencia , Neoplasias Intestinales/dietoterapia , Neoplasias Intestinales/metabolismo , Linfoma/dietoterapia , Linfoma/metabolismo , Serina/deficiencia , Animales , Antioxidantes/metabolismo , Biguanidas/farmacología , Línea Celular Tumoral , Dieta , Modelos Animales de Enfermedad , Femenino , Privación de Alimentos , Glicina/metabolismo , Humanos , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Linfoma/patología , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estado Nutricional , Fosforilación Oxidativa/efectos de los fármacos , Neoplasias Pancreáticas/dietoterapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Serina/biosíntesis , Serina/metabolismo , Serina/farmacología , Tasa de Supervivencia
7.
Mol Cell ; 60(2): 195-207, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26474064

RESUMEN

Cancer cells adapt metabolically to proliferate under nutrient limitation. Here we used combined transcriptional-metabolomic network analysis to identify metabolic pathways that support glucose-independent tumor cell proliferation. We found that glucose deprivation stimulated re-wiring of the tricarboxylic acid (TCA) cycle and early steps of gluconeogenesis to promote glucose-independent cell proliferation. Glucose limitation promoted the production of phosphoenolpyruvate (PEP) from glutamine via the activity of mitochondrial PEP-carboxykinase (PCK2). Under these conditions, glutamine-derived PEP was used to fuel biosynthetic pathways normally sustained by glucose, including serine and purine biosynthesis. PCK2 expression was required to maintain tumor cell proliferation under limited-glucose conditions in vitro and tumor growth in vivo. Elevated PCK2 expression is observed in several human tumor types and enriched in tumor tissue from non-small-cell lung cancer (NSCLC) patients. Our results define a role for PCK2 in cancer cell metabolic reprogramming that promotes glucose-independent cell growth and metabolic stress resistance in human tumors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación Neoplásica de la Expresión Génica , Gluconeogénesis/genética , Neoplasias Pulmonares/metabolismo , Neoplasias/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Adaptación Fisiológica/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Ciclo del Ácido Cítrico/genética , Glucosa/deficiencia , Glutamina/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metabolómica , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/patología , Fosfoenolpiruvato/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Purinas/biosíntesis , Ácido Pirúvico/metabolismo , Serina/biosíntesis
8.
J Cell Sci ; 133(5)2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32144194

RESUMEN

The importance of cancer-cell-autonomous functions of the tumour suppressor p53 (encoded by TP53) has been established in many studies, but it is now clear that the p53 status of the cancer cell also has a profound impact on the immune response. Loss or mutation of p53 in cancers can affect the recruitment and activity of myeloid and T cells, allowing immune evasion and promoting cancer progression. p53 can also function in immune cells, resulting in various outcomes that can impede or support tumour development. Understanding the role of p53 in tumour and immune cells will help in the development of therapeutic approaches that can harness the differential p53 status of cancers compared with most normal tissue.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Humanos , Inmunidad , Mutación , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética
9.
Proc Natl Acad Sci U S A ; 116(18): 8975-8984, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30988188

RESUMEN

T cell help in humoral immunity includes interactions of B cells with activated extrafollicular CD4+ and follicular T helper (Tfh) cells. Each can promote antibody responses but Tfh cells play critical roles during germinal center (GC) reactions. After restimulation of their antigen receptor (TCR) by B cells, helper T cells act on B cells via CD40 ligand and secreted cytokines that guide Ig class switching. Hypoxia is a normal feature of GC, raising questions about molecular mechanisms governing the relationship between hypoxia response mechanisms and T cell help to antibody responses. Hypoxia-inducible factors (HIF) are prominent among mechanisms that mediate cellular responses to limited oxygen but also are induced by lymphocyte activation. We now show that loss of HIF-1α or of both HIF-1α and HIF-2α in CD4+ T cells compromised essential functions in help during antibody responses. HIF-1α depletion from CD4+ T cells reduced frequencies of antigen-specific GC B cells, Tfh cells, and overall antigen-specific Ab after immunization with sheep red blood cells. Compound deficiency of HIF-1α and HIF-2α led to humoral defects after hapten-carrier immunization. Further, HIF promoted CD40L expression while restraining the FoxP3-positive CD4+ cells in the CXCR5+ follicular regulatory population. Glycolysis increases T helper cytokine expression, and HIF promoted glycolysis in T helper cells via TCR or cytokine stimulation, as well as their production of cytokines that direct antibody class switching. Indeed, IFN-γ elaboration by HIF-deficient in vivo-generated Tfh cells was impaired. Collectively, the results indicate that HIF transcription factors are vital components of the mechanisms of help during humoral responses.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Hipoxia de la Célula/inmunología , Hipoxia de la Célula/fisiología , Citocinas/metabolismo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Hipoxia/metabolismo , Inmunidad Humoral , Inmunización , Activación de Linfocitos/inmunología , Activación de Linfocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CXCR5/metabolismo , Ovinos , Linfocitos T Colaboradores-Inductores/inmunología
10.
Proc Natl Acad Sci U S A ; 115(10): E2202-E2209, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463754

RESUMEN

The translation of mRNAs into proteins serves as a critical regulatory event in gene expression. In the context of cancer, deregulated translation is a hallmark of transformation, promoting the proliferation, survival, and metastatic capabilities of cancer cells. The best-studied factor involved in the translational control of cancer is the eukaryotic translation initiation factor 4E (eIF4E). We and others have shown that eIF4E availability and phosphorylation promote metastasis in mouse models of breast cancer by selectively augmenting the translation of mRNAs involved in invasion and metastasis. However, the impact of translational control in cell types within the tumor microenvironment (TME) is unknown. Here, we demonstrate that regulatory events affecting translation in cells of the TME impact cancer progression. Mice bearing a mutation in the phosphorylation site of eIF4E (S209A) in cells comprising the TME are resistant to the formation of lung metastases in a syngeneic mammary tumor model. This is associated with reduced survival of prometastatic neutrophils due to decreased expression of the antiapoptotic proteins BCL2 and MCL1. Furthermore, we demonstrate that pharmacological inhibition of eIF4E phosphorylation prevents metastatic progression in vivo, supporting the development of phosphorylation inhibitors for clinical use.


Asunto(s)
Neoplasias de la Mama/patología , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neutrófilos/metabolismo , Biosíntesis de Proteínas , Microambiente Tumoral , Secuencias de Aminoácidos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Factor 4E Eucariótico de Iniciación/química , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Metástasis de la Neoplasia , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Immunol Rev ; 249(1): 59-71, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22889215

RESUMEN

When T cells encounter foreign antigen and appropriate costimulatory signals from professional antigen-presenting cells (APCs), they initiate a coordinated program of rapid proliferation and differentiation, leading to the development of activated T cells with specific effector functions tailored toward pathogen clearance or control. One of the fundamental programs that underpin T-cell proliferation and function is the regulation of cellular metabolism. Recent efforts to identify the signal transduction pathways that regulate T-cell metabolism have led to the identification of liver kinase B1 (LKB1) and AMP-activated protein kinase (AMPK) as key regulators of T-cell metabolism. LKB1 and AMPK are part of an evolutionarily conserved signal transduction pathway that monitors cellular energy status. AMPK senses bioenergetic fluctuations in cells and works in concert with LKB1 to maintain cellular energy homeostasis by promoting catabolic pathways of ATP production and limiting processes that consume ATP. Recent data indicate that LKB1 and AMPK can influence diverse aspects of T-cell biology beyond metabolism, including T-cell development, peripheral T-cell homeostasis, and T-cell effector function. In this review, we focus on the regulation of lymphocyte metabolism by this energy-sensing pathway and discuss its influence on T-cell function.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antígenos/inmunología , Activación de Linfocitos , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Diferenciación Celular , Proliferación Celular , Metabolismo Energético , Humanos , Biosíntesis de Proteínas , Transducción de Señal , Transcripción Genética
12.
J Immunol ; 189(9): 4349-60, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23028053

RESUMEN

Stomatin-like protein 2 (SLP-2) is a mostly mitochondrial protein that regulates mitochondrial biogenesis and function and modulates T cell activation. To determine the mechanism of action of SLP-2, we generated T cell-specific SLP-2-deficient mice. These mice had normal numbers of thymocytes and T cells in the periphery. However, conventional SLP-2-deficient T cells had a posttranscriptional defect in IL-2 production in response to TCR ligation, and this translated into reduced CD4(+) T cell responses. SLP-2 deficiency was associated with impaired cardiolipin compartmentalization in mitochondrial membranes, decreased levels of the NADH dehydrogenase (ubiquinone) iron-sulfur protein 3, NADH dehydrogenase (ubiquinone) 1ß subcomplex subunit 8, and NADH dehydrogenase (ubiquinone) 1α subcomplex subunit 9 of respiratory complex I, and decreased activity of this complex as well as of complex II plus III of the respiratory chain. In addition, SLP-2-deficient T cells showed a significant increase in uncoupled mitochondrial respiration and a greater reliance on glycolysis. Based on these results, we propose that SLP-2 organizes the mitochondrial membrane compartmentalization of cardiolipin, which is required for optimal assembly and function of respiratory chain complexes. This function, in T cells, helps to ensure proper metabolic response during activation.


Asunto(s)
Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Proteínas Sanguíneas/fisiología , Linfocitos T CD4-Positivos/patología , Cardiolipinas/inmunología , Cardiolipinas/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedades Mitocondriales/metabolismo , Membranas Mitocondriales/inmunología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Subgrupos de Linfocitos T/patología
13.
J Immunol ; 187(8): 4187-98, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21930968

RESUMEN

T cell activation leads to engagement of cellular metabolic pathways necessary to support cell proliferation and function. However, our understanding of the signal transduction pathways that regulate metabolism and their impact on T cell function remains limited. The liver kinase B1 (LKB1) is a serine/threonine kinase that links cellular metabolism with cell growth and proliferation. In this study, we demonstrate that LKB1 is a critical regulator of T cell development, viability, activation, and metabolism. T cell-specific ablation of the gene that encodes LKB1 resulted in blocked thymocyte development and a reduction in peripheral T cells. LKB1-deficient T cells exhibited defects in cell proliferation and viability and altered glycolytic and lipid metabolism. Interestingly, loss of LKB1 promoted increased T cell activation and inflammatory cytokine production by both CD4(+) and CD8(+) T cells. Activation of the AMP-activated protein kinase (AMPK) was decreased in LKB1-deficient T cells. AMPK was found to mediate a subset of LKB1 functions in T lymphocytes, as mice lacking the α1 subunit of AMPK displayed similar defects in T cell activation, metabolism, and inflammatory cytokine production, but normal T cell development and peripheral T cell homeostasis. LKB1- and AMPKα1-deficient T cells each displayed elevated mammalian target of rapamycin complex 1 signaling and IFN-γ production that could be reversed by rapamycin treatment. Our data highlight a central role for LKB1 in T cell activation, viability, and metabolism and suggest that LKB1-AMPK signaling negatively regulates T cell effector function through regulation of mammalian target of rapamycin activity.


Asunto(s)
Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Proliferación Celular , Separación Celular , Supervivencia Celular/inmunología , Citometría de Flujo , Homeostasis/inmunología , Immunoblotting , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Linfocitos T/citología
14.
Sci Adv ; 9(36): eadh2023, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37672588

RESUMEN

Previous studies have revealed a role for proline metabolism in supporting cancer development and metastasis. In this study, we show that many cancer cells respond to loss of attachment by accumulating and secreting proline. Detached cells display reduced proliferation accompanied by a general decrease in overall protein production and de novo amino acid synthesis compared to attached cells. However, proline synthesis was maintained under detached conditions. Furthermore, while overall proline incorporation into proteins was lower in detached cells compared to other amino acids, there was an increased production of the proline-rich protein collagen. The increased excretion of proline from detached cells was also shown to be used by macrophages, an abundant and important component of the tumor microenvironment. Our study suggests that detachment induced accumulation and secretion of proline may contribute to tumor progression by supporting increased production of extracellular matrix and providing proline to surrounding stromal cells.


Asunto(s)
Neoplasias , Prolina , Aminoácidos , Transporte Biológico , Matriz Extracelular , Macrófagos
15.
Cell Rep ; 42(6): 112562, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37245210

RESUMEN

Mitochondrial 10-formyltetrahydrofolate (10-formyl-THF) is utilized by three mitochondrial enzymes to produce formate for nucleotide synthesis, NADPH for antioxidant defense, and formyl-methionine (fMet) to initiate mitochondrial mRNA translation. One of these enzymes-aldehyde dehydrogenase 1 family member 2 (ALDH1L2)-produces NADPH by catabolizing 10-formyl-THF into CO2 and THF. Using breast cancer cell lines, we show that reduction of ALDH1L2 expression increases ROS levels and the production of both formate and fMet. Both depletion of ALDH1L2 and direct exposure to formate result in enhanced cancer cell migration that is dependent on the expression of the formyl-peptide receptor (FPR). In various tumor models, increased ALDH1L2 expression lowers formate and fMet accumulation and limits metastatic capacity, while human breast cancer samples show a consistent reduction of ALDH1L2 expression in metastases. Together, our data suggest that loss of ALDH1L2 can support metastatic progression by promoting formate and fMet production, resulting in enhanced FPR-dependent signaling.


Asunto(s)
Neoplasias de la Mama , Formiatos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Femenino , Humanos , Neoplasias de la Mama/metabolismo , Formiatos/metabolismo , Metionina , NADP , Especies Reactivas de Oxígeno , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo
16.
Cell Metab ; 35(7): 1132-1146.e9, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37230079

RESUMEN

Augmented T cell function leading to host damage in autoimmunity is supported by metabolic dysregulation, making targeting immunometabolism an attractive therapeutic avenue. Canagliflozin, a type 2 diabetes drug, is a sodium glucose co-transporter 2 (SGLT2) inhibitor with known off-target effects on glutamate dehydrogenase and complex I. However, the effects of SGLT2 inhibitors on human T cell function have not been extensively explored. Here, we show that canagliflozin-treated T cells are compromised in their ability to activate, proliferate, and initiate effector functions. Canagliflozin inhibits T cell receptor signaling, impacting on ERK and mTORC1 activity, concomitantly associated with reduced c-Myc. Compromised c-Myc levels were encapsulated by a failure to engage translational machinery resulting in impaired metabolic protein and solute carrier production among others. Importantly, canagliflozin-treated T cells derived from patients with autoimmune disorders impaired their effector function. Taken together, our work highlights a potential therapeutic avenue for repurposing canagliflozin as an intervention for T cell-mediated autoimmunity.


Asunto(s)
Enfermedades Autoinmunes , Diabetes Mellitus Tipo 2 , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Canagliflozina/farmacología , Canagliflozina/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Autoinmunidad , Linfocitos T , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Hipoglucemiantes/farmacología
17.
Blood ; 115(23): 4742-9, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20351312

RESUMEN

Dendritic cells (DCs) are key regulators of innate and acquired immunity. The maturation of DCs is directed by signal transduction events downstream of toll-like receptors (TLRs) and other pattern recognition receptors. Here, we demonstrate that, in mouse DCs, TLR agonists stimulate a profound metabolic transition to aerobic glycolysis, similar to the Warburg metabolism displayed by cancer cells. This metabolic switch depends on the phosphatidyl inositol 3'-kinase/Akt pathway, is antagonized by the adenosine monophosphate (AMP)-activated protein kinase (AMPK), and is required for DC maturation. The metabolic switch induced by DC activation is antagonized by the antiinflammatory cytokine interleukin-10. Our data pinpoint TLR-mediated metabolic conversion as essential for DC maturation and function and reveal it as a potential target for intervention in the control of excessive inflammation and inappropriately regulated immune responses.


Asunto(s)
Células Dendríticas/inmunología , Glucólisis/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Animales , Células Dendríticas/metabolismo , Glucólisis/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/metabolismo , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Toll-Like/antagonistas & inhibidores , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
18.
Front Immunol ; 12: 637960, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868263

RESUMEN

Regulatory T cells (Tregs) are essential for mitigating inflammation. Tregs are found in nearly every tissue and play either beneficial or harmful roles in the host. The availability of various nutrients can either enhance or impair Treg function. Mitochondrial oxidative metabolism plays a major role in supporting Treg differentiation and fitness. While Tregs rely heavily on oxidation of fatty acids to support mitochondrial activity, they have found ways to adapt to different tissue types, such as tumors, to survive in competitive environments. In addition, metabolic by-products from commensal organisms in the gut also have a profound impact on Treg differentiation. In this review, we will focus on the core metabolic pathways engaged in Tregs, especially in the context of tissue nutrient environments, and how they can affect Treg function, stability and differentiation.


Asunto(s)
Inflamación/inmunología , Microbiota/fisiología , Mitocondrias/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular , Humanos , Inmunomodulación , Nutrientes , Oxidación-Reducción
19.
Nat Commun ; 12(1): 1209, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33619282

RESUMEN

Fructose intake has increased substantially throughout the developed world and is associated with obesity, type 2 diabetes and non-alcoholic fatty liver disease. Currently, our understanding of the metabolic and mechanistic implications for immune cells, such as monocytes and macrophages, exposed to elevated levels of dietary fructose is limited. Here, we show that fructose reprograms cellular metabolic pathways to favour glutaminolysis and oxidative metabolism, which are required to support increased inflammatory cytokine production in both LPS-treated human monocytes and mouse macrophages. A fructose-dependent increase in mTORC1 activity drives translation of pro-inflammatory cytokines in response to LPS. LPS-stimulated monocytes treated with fructose rely heavily on oxidative metabolism and have reduced flexibility in response to both glycolytic and mitochondrial inhibition, suggesting glycolysis and oxidative metabolism are inextricably coupled in these cells. The physiological implications of fructose exposure are demonstrated in a model of LPS-induced systemic inflammation, with mice exposed to fructose having increased levels of circulating IL-1ß after LPS challenge. Taken together, our work underpins a pro-inflammatory role for dietary fructose in LPS-stimulated mononuclear phagocytes which occurs at the expense of metabolic flexibility.


Asunto(s)
Fructosa/farmacología , Glutamina/metabolismo , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/toxicidad , Ácidos/metabolismo , Animales , Ciclo del Ácido Cítrico/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Glucosa/farmacología , Glucólisis/efectos de los fármacos , Marcaje Isotópico , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Análisis de Flujos Metabólicos , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Fenotipo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
20.
Cell Rep ; 30(2): 481-496.e6, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31940491

RESUMEN

Loss of p53 function contributes to the development of many cancers. While cell-autonomous consequences of p53 mutation have been studied extensively, the role of p53 in regulating the anti-tumor immune response is still poorly understood. Here, we show that loss of p53 in cancer cells modulates the tumor-immune landscape to circumvent immune destruction. Deletion of p53 promotes the recruitment and instruction of suppressive myeloid CD11b+ cells, in part through increased expression of CXCR3/CCR2-associated chemokines and macrophage colony-stimulating factor (M-CSF), and attenuates the CD4+ T helper 1 (Th1) and CD8+ T cell responses in vivo. p53-null tumors also show an accumulation of suppressive regulatory T (Treg) cells. Finally, we show that two key drivers of tumorigenesis, activation of KRAS and deletion of p53, cooperate to promote immune tolerance.


Asunto(s)
Células Mieloides/metabolismo , Linfocitos T Reguladores/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA